Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Drug Metabolism & Disposition
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Drug Metabolism & Disposition

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit dmd on Facebook
  • Follow dmd on Twitter
  • Follow ASPET on LinkedIn
Research Article50th Anniversary Celebration Collection—Minireview

Species Specificity and Selection of Models for Drug Oxidations Mediated by Polymorphic Human Enzymes

Hiroshi Yamazaki and Makiko Shimizu
Drug Metabolism and Disposition January 2023, 51 (1) 123-129; DOI: https://doi.org/10.1124/dmd.121.000742
Hiroshi Yamazaki
Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Makiko Shimizu
Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Visual Overview

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Abstract

Many drug oxygenations are mainly mediated by polymorphic cytochromes P450 (P450s) and also by flavin-containing monooxygenases (FMOs). More than 50 years of research on P450/FMO-mediated drug oxygenations have clarified their catalytic roles. The natural product coumarin causes hepatotoxicity in rats via the reactive coumarin 3,4-epoxide, a reaction catalyzed by P450 1A2; however, coumarin undergoes rapid 7-hydroxylation by polymorphic P450 2A6 in humans. The primary oxidation product of the teratogen thalidomide in rats is deactivated 5′-hydroxythalidomide plus sulfate and glucuronide conjugates; however, similar 5′-hydroxythalidomide and 5-hydroxythalidomide are formed in rabbits in vivo. Thalidomide causes human P450 3A enzyme induction in liver (and placenta) and is also activated in vitro and in vivo by P450 3A through the primary human metabolite 5-hydroxythalidomide (leading to conjugation with glutathione/nonspecific proteins). Species differences exist in terms of drug metabolism in rodents and humans, and such differences can be very important when determining the contributions of individual enzymes. The approaches used for investigating the roles of human P450 and FMO enzymes in understanding drug oxidations and clinical therapy have not yet reached maturity and still require further development.

SIGNIFICANCE STATEMENT Drug oxidations in animals and humans mediated by P450s and FMOs are important for understanding the pharmacological properties of drugs, such as the species-dependent teratogenicity of the reactive metabolites of thalidomide and the metabolism of food-derived odorous trimethylamine to non-odorous (but proatherogenic) trimethylamine N-oxide. Recognized differences exist in terms of drug metabolism between rodents, non-human primates, and humans, and such differences are important when determining individual liver enzyme contributions with substrates in in vitro and in vivo systems.

Introduction

Many drug metabolism reactions are mediated by cytochromes P450 (P450s, EC 1.14.14.1). The first article on P450 was published in 1962 (Omura and Sato, 1962). In 2022, we celebrate 60 years of research on the many forms of P450 that exist in animals and humans. P450 research has come a long way from early studies in the 1960s to the current era of personalized medicine in individual patients. Drug oxidation reactions are also catalyzed by several non-P450 enzymes in humans (Rendic and Guengerich, 2015, Rendic et al., 2022), e.g., flavin-containing monooxygenases (FMOs) [previously referred to as liver microsomal mixed-function amine oxidases (Masters and Ziegler, 1971) or flavin adenine dinucleotide-containing monooxygenase (Poulsen and Ziegler, 1979)]. Both P450s and FMOs require NADPH and oxygen for the oxygenation of substrates (Ziegler, 1988). FMOs (EC 1.14.13.8) are a family of NADPH-dependent enzymes that oxygenate a range of heteroatom-containing substances (Krueger and Williams, 2005; Cashman and Zhang, 2006; Phillips and Shephard, 2020), including benzydamine and trimethylamine. Many drug-drug interactions are pharmacokinetic in nature and caused by modifications of drug metabolism, generally through enzyme inhibition or enzyme induction (Rendic and Guengerich, 2010). In vitro determinations of the roles of the enzymes primarily responsible for specific drug oxidation reactions generally use human liver preparations and constitute a key method for estimating and understanding a variety of drug interactions (Brown et al., 2005; Shimizu et al., 2021b). To address the inhibitory potential of a drug, it is important to determine inhibition parameters in in vitro systems (Ito et al., 1998) and to elucidate the individual enzyme contributions in vitro (i.e., fraction metabolized by each enzyme, fm) to substrate oxidations in livers (Youdim et al., 2008).

Rodents are often used as animal models in drug development; however, it is well known that species differences exist in terms of drug metabolism between rodent and human cytochrome P450s (Kazuki et al., 2016; Yamazaki et al., 2016b). The relevance and limitations of animal models used in non-clinical safety assessments of investigational products should be carefully considered. For example, coumarin (Fig. 1A), a naturally occurring food flavoring (Lake, 1999; Abraham et al., 2010), has been reported to cause hepatotoxicity in rats via the reactive metabolite coumarin 3,4-epoxide, which is important because coumarin is a structural base for a family of anticoagulant rodenticides (Vassallo et al., 2003; Rietjens et al., 2010). Coumarin has also been used as a medicine in humans and has a tolerable daily intake of 0.1 mg/kg of body weight (Abraham et al., 2010). Apparent species differences should be noted between the rapid coumarin 7-hydroxylation seen in humans (Yamazaki et al., 1994; Kiyotani et al., 2003) and the slow coumarin 7-hydroxylation in rats (Murayama and Yamazaki, 2021). Thalidomide is another substrate with notable species differences in terms of metabolism. Thalidomide is teratogenic in non-human primates and rabbits (Calabrese and Resztak, 1998) but not in rodents (Kim and Scialli, 2011). Among a range of animal species, thalidomide is differentially oxidized to either 5-hydroxythalidomide or 5′-hydroxythalidomide (Nishiyama et al., 2015) (Fig. 1B).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Oxidation of coumarin to o-hydroxyphenylacetic acid and 7-hydroxycoumarin (A) (Miura et al., 2021a) and of thalidomide to 5′-hydroxythalidomide and 5-hydroxythalidomide (B) (Kuwagata et al., 2021; Miura et al., 2021b).

Applications of P450 research in drug metabolism studies continue to expand and develop. Further, interest has also been focused on a member of the flavin-containing monooxygenase (FMO) family, namely FMO3, because it is responsible for N- and S-oxygenations of various drugs, and FMO5 has also recently been shown to catalyze Bäyer-Villiger reaction, leading to the formation of esters (Rendic et al., 2022). FMO3 is of particular interest because genetic polymorphism in the human FMO3 gene can affect its catalytic efficiency with respect to typical medicinal substrates (Catucci et al., 2012; Nakamaru et al., 2014) and thereby lead to drug interactions (Yamazaki and Shimizu, 2013). Phenotype–gene analyses have revealed impaired FMO3 variants associated with the metabolic disorder trimethylaminuria, a condition that causes an unpleasant fishy smell (Humbert et al., 1970). FMO3 extensively catalyzes trimethylamine N-oxygenation to produce non-odorous (Shephard et al., 2012) but proatherogenic (Hartiala et al., 2014) trimethylamine N-oxide.

Research on P450s in non-human primates (Uno et al., 2022c) and in animal models with introduced human P450 (CYP) genes or transplanted with human liver cells (Uehara et al., 2022) has extended into different fields, from molecular to in vivo research. The present minireview summarizes recent findings on polymorphic P450- and FMO-mediated oxidation and bioactivation reactions. This research has furthered our understanding of the pharmacological properties of drugs such as teratogenic thalidomide and food-derived trimethylamine and provides important fundamental information for future investigations of species specificity and selection of models for drug oxidations mediated by polymorphic human enzymes.

Brief Historical Perspective

The extensive contributions of scientists throughout the world to the field of P450 research over past six decades is noteworthy. The success of P450 research has had implications in disciplines such as drug-drug interactions and pharmacogenetics.

Because thalidomide was previously considered to undergo very little metabolism by the P450 system, drug interactions between thalidomide and hormonal contraceptives have been considered unlikely (Trapnell et al., 1998; Teo et al., 2000). However, this was recently shown not to be the case. The ubiquitously expressed E3 ligase protein cereblon is reportedly a common direct protein target explaining the antiproliferative activities of thalidomide, lenalidomide, and pomalidomide (Ito et al., 2010; Lopez-Girona et al., 2012); however, this target also exists in rodents, which show no thalidomide teratogenicity (Kim and Scialli, 2011). Its teratogenic effects notwithstanding, thalidomide was subsequently approved for the clinical treatment of multiple myeloma (Palumbo et al., 2008; Nakamura et al., 2013), at which point the effects of thalidomide on P450 activities were investigated again in more detail. Thalidomide enhanced P450 3A4/5-dependent midazolam hydroxylation and cyclosporine A clearance at clinically relevant concentrations (Okada et al., 2009) and induced human P450 3A enzymes through the pregnane X receptor (PXR) (Murayama et al., 2014). Thalidomide was shown to be hydroxylated by human liver microsomal P450 3A4/5 (Chowdhury et al., 2010). Thalidomide is metabolized by two major pathways (Fig. 1B) by various P450s, as previously investigated (Ando et al., 2002; Lu et al., 2004). Furthermore, human P450 3A4 and 3A5 also oxidize thalidomide to form 5-hydroxy and dihydroxy metabolites (Chowdhury et al., 2010; Yamazaki et al., 2011; Yamazaki et al., 2012). The secondary oxidation of 5-hydoxythalidomide is faster than the primary thalidomide 5-hydoxylation mediated by recombinant human P450 3A4/5 in in vitro systems (Chowdhury et al., 2014; Yamazaki et al., 2016a). The second oxidation step in the human P450 3A4 pathway generates a reactive intermediate, possibly an arene oxide, as was suggested earlier (Gordon et al., 1981). Such intermediates can be trapped by to result in glutathione adducts (Yamazaki et al., 2016a) that have been confirmed in vivo in humanized-liver mouse models after oral doses of 100 mg/kg of thalidomide (Yamazaki et al., 2013). Although five control mice had no apparent adverse effects after an oral dose of 270 mg/kg (∼1 mmol) of thalidomide/kg, an oral LD50 of 270 mg/kg was observed for humanized-liver mice (two of five animals died after 2 days) (Miura et al., 2021b). Differences in species susceptibility to thalidomide teratogenicity may result from differences in the primary biotransformation of the compound by drug-metabolizing enzymes.

One historical perspective on the catalytic mechanism of P450 involves electron transport (Fig. 2A), mediated by a multicomponent monooxygenase system based on NADPH (Yamazaki et al., 1996; Yamazaki et al., 2001). Microsomal P450s receive electrons from NADPH-cytochrome P450 reductase. The catalytic cycle of P450 involves the activation of molecular oxygen to a reactive form (Fig. 2A). In contrast, FMOs are characterized by a stable 4a-flavin hydroperoxide intermediate capable of oxygenating both nucleophiles and electrophiles in its catalytic cycle (Fig. 2B), formed even in the absence of a substrate (Jones and Ballou, 1986). The initiation of drug oxidation reactions is generally done with the addition of NADPH when investigating typical P450-mediated reactions. However, with FMOs, the absence of NADPH in the preincubation period decreases FMO-mediated drug oxygenation activities (Taniguchi-Takizawa et al., 2015). NADPH stabilizes of flavin adenine dinucleotide in FMO enzymes, which have a higher pH optimum (8.4) (Nagashima et al., 2009) in the FMO catalytic cycle.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Catalytic cycles of P450s (A) and FMOs (B). As indicated in the catalytic cycle, ferrous P450 can bind substrate (Yamazaki et al., 1996; Yamazaki et al., 2001), whereas for FMOs, only in the third step is the flavin hydroperoxide intermediate capable of oxygenating substrates (Ziegler, 2002).

Historical findings in P450 research should not be neglected when planning new studies. For example, human drug-metabolizing P450s (in phospholipid membranes) require an additional protein, NADPH-cytochrome P450 reductase, in an environment with appropriate ionic strength to catalyze aerobic reactions in the presence of NADPH (Yamazaki et al., 1995). Because benzydamine N-oxygenation and N-demethylation are mediated by FMO1/3 and P450 2D6/3A4, respectively, benzydamine can serve as a suitable probe substrate for both FMOs and P450s for human drug interaction studies. Dimethyl sulfoxide is a widely used organic solvent in biochemical analysis (Hickman et al., 1998; Busby et al., 1999), but a high concentration of dimethyl sulfoxide (more than 4%, v/v) prevents the mutagenic activation of some procarcinogens by P450 enzymes in preincubation procedures, thereby indicating that dimethyl sulfoxide can interfere with P450 metabolism (Mori et al., 1985). P450 2E1 reactions are especially prone to inhibition by solvents, in that most of these are substrates (and a concentration of 1% (v/v) is ∼ 10 mM). Low concentrations of dimethyl sulfoxide (0.1% and 0.5%, v/v) inhibited FMO3 enzyme activities in human liver microsomes and recombinant systems, respectively (Taniguchi-Takizawa et al., 2015). Dimethyl sulfoxide was suggested to be an FMO substrate in humans as a result of in vivo urinary excretion (of dimethyl sulfone (Hucker et al., 1967)) and of its inhibitory effects on in vitro FMO3-dependent oxygenations in human liver microsomes (Taniguchi-Takizawa et al., 2015). The contribution of FMO to the metabolic clearance of new drug candidates could be underestimated under some current high-throughput experimental conditions that employ dimethyl sulfoxide for use with P450 enzymes.

FMOs constitute an enzyme system complementary to the cytochrome P450 (P450) enzyme family, which should be taken into account during drug development (Cashman, 2008). Because FMOs are not readily induced or inhibited, it has been suggested that there may be advantages in designing drugs that are partly metabolized by FMOs, i.e., drugs that are not metabolized exclusively by P450s (Krueger and Williams, 2005; Cashman, 2008). However, the contributions of FMOs to the metabolic elimination of new drug candidates may be underestimated under the usual experimental and physiologic conditions of pH 7.4, which are more suited to P450 enzymes initiated by the addition of NADPH to reaction mixtures containing drug candidates with classic dissociation constants (pKa base) of >8.4 (Taniguchi-Takizawa et al., 2021).

Utility of non-human primates in drug development because of their physiologic and genetic similarities to humans has been separately reviewed with information on the major multiple forms of drug-metabolizing P450 enzymes and FMO3 having generally similar substrate selectivities to those of human P450s and FMO3 (Uno et al., 2016; Uno et al., 2018; Uehara et al., 2020; Uno et al., 2022c).

Some Key Recent Advances

Thalidomide causes human P450 3A enzyme induction/cooperativity in liver (and placenta) and is also activated by P450 3A through its human primary metabolite (5-hydroxythalidomide) in vitro and in vivo (Murayama et al., 2017). Screening for immunomodulatory drugs that possess less teratogenicity/toxicity is a strategic pathway toward the development of new anti-cancer drugs. Lenalidomide (Kim and Schmidt-Wolf, 2015; Chen et al., 2017), a second-generation immunomodulatory analog of thalidomide in which one keto group has been removed (plus the addition of an amino group) has more potent anti-tumor activity than thalidomide (Zeldis et al., 2013). Thalidomide is well distributed to the brain in rodents (with a brain/plasma ratio of 0.89) (Huang et al., 2005) and to cerebrospinal fluid in non-human primates (with a cerebrospinal fluid/plasma ratio of 0.42) (Muscal et al., 2012). In contrast, lenalidomide distribution to the brain in rodents is low (Rozewski et al., 2012). These facts notwithstanding, a clinical response to lenalidomide has been observed in cerebrospinal tumors (Warren et al., 2011). In contrast to the metabolites identified for the first-generation drug thalidomide and the second-generation drug pomalidomide (Shimizu et al., 2017), no significant hydroxylated metabolites of lenalidomide (Murayama et al., 2018a) have been detected in in vitro or in vivo assays (Chowdhury et al., 2014).

Non-human primates, cynomolgus monkeys and common marmosets, are generally expected to show similar drug-metabolizing properties to humans. Reported areas under the plasma concentration curves after oral administrations of the five typical human P450 cocktail probes (caffeine, warfarin, omeprazole, metoprolol, and midazolam) (Turpault et al., 2009) were generally similar among humans, monkeys, and marmosets after normalization by doses per body weights (Koyanagi et al., 2015; Toda et al., 2018). However, caffeine is metabolized by human P450 1A2 to paraxanthine (N3-demethylation) (Turpault et al., 2009), by monkey P450 2C9 to theophylline (N7-demethylation) (Utoh et al., 2016), and by marmoset P450 1A2 and 3A4 to N3-demethylated and 8-hydroxylated metabolites, respectively (Uehara et al., 2016). Some attention should be paid to the differences even in non-human primates and humans.

The fraction of victim drug metabolized by an enzyme (fm) in vitro is one of the parameters used to estimate in vivo drug–drug interactions. The fm value is usually determined by comparing the intrinsic clearances of victim drugs for selectively inhibited and uninhibited hepatic microsomes in vitro. Pooled human liver microsomes in which one P450 enzyme has been selectively inactivated by a suitable mechanism-based inhibitor using preincubation methods has been commercially introduced by the pharmaceutical industry (Murayama et al., 2018b; Parmentier et al., 2019), effectively yielding in vitro “knock-outs.” Furafylline, a 1,3,8-trisubstituted xanthine, results in the loss of human P450 1A2 activity (Racha et al., 1998). Coumarin oxidations investigated were 3,4-epoxidation, ultimately leading to the formation of o-hydroxyphenylacetic acid, and 7-hydroxylation in humanized-liver mice treated with furafylline, a mechanism-based inhibitor of P450 1A2 (Racha et al., 1998). The strategy of using suppressed liver P450 1A2 activities in humanized-liver mice was introduced to elucidate the roles of the enzymes in the in vivo activation of coumarin to its primary reactive 3,4-epoxide (Miura et al., 2021a). Using the mechanism-based inactivation of P450 2C9 by tienilic acid (Hutzler et al., 2009), we could precisely assess the oxidation activities of selectively inactivated human liver microsomes to determine the in vitro contributions of P450 2C9 and 3A enzymes to the metabolism of diclofenac at different ionic strengths of incubation buffers (Miura et al., 2020). We also compared the roles of P450 2C9 and 3A in metabolite formation from S-warfarin in vivo using humanized-liver mice. A modified strategy using the selective in vivo inactivation of liver P450 2C9 in humanized-liver mice was developed to provide important information on the differences between in vitro and in vivo drug metabolite formation from diclofenac, a typical P450 2C9 probe substrate used in in vitro experiments (Miura et al., 2020).

Traditional urinary screening tests for the metabolic capacity of FMO3 in individual humans have also been carried out (Yamazaki and Shimizu, 2007; Yamazaki and Shimizu, 2013). Some updated findings for the metabolic capacity of FMO3 screened on the basis of urine testing of trimethylamine N-oxide levels are shown in Fig. 3. The variability in in vivo activities was primarily attributed to FMO3 variants. Increasing numbers of single-nucleotide substitutions of the human FMO3 gene are being recorded in mega-databases (Shimizu et al., 2019). A series of reliable FMO3 genotyping confirmation methods has been assembled and developed for ∼40 impaired FMO3 variants (Shimizu et al., 2021a). This series of systems should facilitate the easy detection in the clinical setting of FMO3 variants in subjects susceptible to low drug clearances or drug interactions possibly caused by impaired FMO3 function.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Metabolic capacity of FMO3 screened on the basis of urine testing of trimethylamine N-oxide levels. Japanese volunteers with self-reported malodor who responded to an Internet article for screening were investigated, and the data were updated from those of our previous study (Yamazaki and Shimizu, 2007). The frequency of individuals with <40% of wild-type FMO3 metabolic capacity in the self-reported population was 1.5% in this Japanese cohort.

Current Challenges and Knowledge Gaps

The success of research on drug metabolizing enzymes and its application in drug discovery and development has been remarkable. Because most of the major recombinant human drug-metabolizing P450s are now commercially available as enzyme sources, it might be thought that adequate procedures for determining the roles of human P450 enzymes in new drug oxidations have been developed and have reached maturity, but this is not necessarily the case. For example, the P450 forms responsible for hydroxylation of the antihistamine drug ebastine could not be previously identified using the recombinant P450 enzymes commercially available. Using more traditional inhibition studies, human intestinal P450 2J2 and 4F12 were found to metabolize endogenous substrates and also to be involved in the metabolism of ebastine (Hashizume et al., 2002). Information from package inserts for some clinically important medicines, such as the antibiotic linezolid, indicates that no roles of major human P450 enzymes have been identified in liver microsomal drug oxidations, although those studies did not rule out the role of currently uncharacterized P450s (Wynalda et al., 2000). Similarly, recent findings from in vitro assays for linezolid have shown contribution of P450 2J2 (Obach, 2022), an enzyme not generally included in testing at the preclinical drug development stage.

The relative levels of P450 expressions in livers show similarities in humans (Shimada et al., 1994), monkeys (Uehara et al., 2011), and marmosets (Shimizu et al., 2014). Although it should be noted that expression levels are not always comparable between mRNA and protein levels, P450 3A expression is most abundant and P450 2C expression is moderately abundant in human and monkey livers in terms of protein levels and in marmoset livers in terms of mRNA levels. In addition, monkey P450 2As (Uehara et al., 2011) and marmoset P450 2Ds (Shimizu et al., 2014) are more abundant, but monkey and marmoset P450 1As are less abundant compared with their human P450 counterparts (Shimada et al., 1994).

Most major drug metabolizing enzymes are known to be polymorphic, or strictly speaking “variant” (polymorphism is defined as an incidence ≥1%). Although the molecular structure of the P450 2C9.2 variant has been solved using X-ray crystallography (Parikh et al., 2020), it is not realistic to evaluate the functional activity of minor variant forms of P450 enzymes in in vitro systems, only some major ones (e.g., 2C9.2 and 2C9.3). Examples of application of knowledge of P450 variants to clinical practice are still few. Digital simulations using physiologically based pharmacokinetic models may be useful additions to the estimations of drug concentrations in human blood in advance of pre-clinical and clinical studies (Ota et al., 2019; Notsu et al., 2020). Pharmacokinetic modeling of in vivo drug concentrations could serve as a useful guide when assessing and setting treatments for clinical patients with polymorphic variations of major drug-metabolizing enzymes (Ota et al., 2019; Notsu et al., 2020). A major problem is that coding-region variants have different effects with individual substrates (Yamazaki et al., 1998).

Perspective on Future Directions

The in vitro methods used to assess the inhibitory potential of drugs and the individual contributions of enzymes to substrate oxidation in human liver vary in different laboratories. Consequently, more standardized methods are required. The application of human liver microsomes with specifically inactivated P450 enzymes can provide information about the development of inactivated-P450 human hepatocytes before transplantation into immunodeficient mice, thereby creating a standard animal model for investigating the contributions of specific P450s in humans (Miura et al., 2020; Miura et al., 2021a). However, these will not be stable mouse lines, in that they are not genetically altered.

To facilitate extrapolation of the in vivo pharmacokinetics of drugs from data obtained in non-human primates to humans, polymorphic drug oxygenation enzymes were investigated in monkeys and marmosets (Uno et al., 2016; Uno et al., 2018; Uehara et al., 2020; Uno et al., 2022c). Veterinary applications for dogs, pigs, and other animals can be anticipated not only for human model animals but also for pets and domestic farm animals as clinical targets with human medicines. For instance, comparisons of human enzymes with those of animal species used in preclinical studies have shown that dogs exhibited hepatic metabolic activities toward human P450 3A substrates erythromycin and nifedipine at levels similar to those of humans, whereas cynomolgus macaques, guinea pigs, and rat enzymes showed substantially greater activities (Shimada et al., 1997). A newly identified dog P450 3A98, a testosterone 6β- and estradiol 16α-hydroxylase, was abundantly expressed in small intestine and is likely the major P450 3A enzyme in the small intestine in combination with dog liver-specific P450 3A12 (Uno et al., 2022a). Roles of new dog P450 2J2, cat P450 2J2, and pig P450 2J33, 2J34, and 2J35 were recently investigated (Uno et al., 2022b). Liver microsomal FMO1/3 were functionally characterized in monkeys, marmosets, dogs, minipigs, and rats in vivo and/or in vitro (Yamazaki et al., 2014; Taniguchi-Takizawa et al., 2015; Uehara et al., 2017; Shimizu et al., 2020; Uno et al., 2022d).

Because human P450 2A6 mediates nicotine oxidation and also the metabolic activation of tobacco-related procarcinogens (Yamazaki et al., 1992; Shimada et al., 2016), it has been postulated that polymorphic P450 2A6 is involved in tumor initiation (Fujieda et al., 2004). Human biomonitoring studies have suggested that heterocyclic amines and N-nitrosamines in meat-derived products may play important roles in colorectal carcinogenesis (Wu et al., 2006). A chemopreventive effect of low-dose aspirin against colorectal tumors was previously found in patients, especially in non-smokers, who had been treated for colorectal adenomas or adenocarcinomas or who had more than 100 adenomatous polyps (Ishikawa et al., 2014; Ishikawa et al., 2021). P450 2A6 is not the enzyme responsible for aspirin metabolism, in which the major human P450 involved in both 3- and 5-hydroxylation of salicylic acid is P450 2E1 (Bojic et al., 2015). However, the CYP2A6 wild-type allele could be a candidate biomarker for reduced chemopreventive effects of daily aspirin, due to unknown mechanisms, and this hypothesis could be applicable to future personalized treatments (Yamazaki et al., 2021). The methods used to investigate the roles of human P450 enzymes in drug oxidations and clinical treatments have not yet reached maturity and require further development.

Conclusions

This minireview was developed for the special section of drug metabolism: a half-century plus of progress, continued need, and new opportunities. P450 research has come a long way from early studies in the 1960s to the current era of personalized medicine (especially with respect to drug metabolism mediated by polymorphic P450s) in individual patients, including children and the elderly. P450s have been the focus of attention in many pharmaceutical companies and industrial applications. Research on P450s using non-human primates and animal models with introduced human P450 (CYP) genes or with transplanted human hepatocytes has attracted biochemists, pharmacologists, and toxicologists and the scope has broadened from molecular research to in vivo investigations. P450/FMO research developed from early studies with animal liver to now include personalized medicine informed by studies of polymorphic P450s/FMO3 in individual patients, including children. The success of drug oxidation research has had implications in fields, such as herbal medicine, drug-drug interactions, pharmacogenetics, and physiologically based pharmacokinetic modeling. The extensive contributions of scientists throughout the world to the field of drug oxygenation research over a half-century plus should be recognized. The techniques used for investigating the roles of human P450/FMO enzymes in drug oxidations and clinical treatments have not yet reached maturity and require further development. We encourage more young scientists to join the important and exciting world of basic and advanced research on drug oxidation and to both become familiar with traditional techniques and strive to develop new methods and approaches.

Acknowledgments

The authors thank Professor Frederick Peter Guengerich, a recipient of the Bernard B. Brodie Award in Drug Metabolism, for his kind and continuous support through the years. The authors also thank David Smallbones for copyediting a draft of this article.

Authorship Contributions

Participated in research design: Yamazaki.

Performed data analysis: Yamazaki, Shimizu.

Wrote or contributed to the writing of the manuscript: Yamazaki.

Footnotes

    • Received October 27, 2021.
    • Accepted June 1, 2022.
  • P450 and FMO research in the authors’ laboratory was supported in part by the Japan Society for the Promotion of Science Grant-in-Aid for Scientific Research [Grant 20K07164].

  • The authors declare that they have no conflicts of interest with respect to the contents of this article.

  • dx.doi.org/10.1124/dmd.121.000742.

Abbreviations

fm
fraction metabolized by each enzyme
FMO
flavin-containing monooxygenase
P450
cytochrome P450
  • Copyright © 2022 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Abraham K,
    2. Wöhrlin F,
    3. Lindtner O,
    4. Heinemeyer G, and
    5. Lampen A
    (2010) Toxicology and risk assessment of coumarin: focus on human data. Mol Nutr Food Res 54:228–239.
    OpenUrlPubMed
  2. ↵
    1. Ando Y,
    2. Fuse E, and
    3. Figg WD
    (2002) Thalidomide metabolism by the CYP2C subfamily. Clin Cancer Res 8:1964–1973.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Bojić M,
    2. Sedgeman CA,
    3. Nagy LD, and
    4. Guengerich FP
    (2015) Aromatic hydroxylation of salicylic acid and aspirin by human cytochromes P450. Eur J Pharm Sci 73:49–56.
    OpenUrl
  4. ↵
    1. Brown HS,
    2. Ito K,
    3. Galetin A, and
    4. Houston JB
    (2005) Prediction of in vivo drug-drug interactions from in vitro data: impact of incorporating parallel pathways of drug elimination and inhibitor absorption rate constant. Br J Clin Pharmacol 60:508–518.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Busby WF Jr.,
    2. Ackermann JM, and
    3. Crespi CL
    (1999) Effect of methanol, ethanol, dimethyl sulfoxide, and acetonitrile on in vitro activities of cDNA-expressed human cytochromes P-450. Drug Metab Dispos 27:246–249.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Calabrese L and
    2. Resztak K
    (1998) Thalidomide revisited: pharmacology and clinical applications. Expert Opin Investig Drugs 7:2043–2060.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Cashman JR
    (2008) Role of flavin-containing monooxygenase in drug development. Expert Opin Drug Metab Toxicol 4:1507–1521.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Cashman JR and
    2. Zhang J
    (2006) Human flavin-containing monooxygenases. Annu Rev Pharmacol Toxicol 46:65–100.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Catucci G,
    2. Gilardi G,
    3. Jeuken L, and
    4. Sadeghi SJ
    (2012) In vitro drug metabolism by C-terminally truncated human flavin-containing monooxygenase 3. Biochem Pharmacol 83:551–558.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Chen N,
    2. Zhou S, and
    3. Palmisano M
    (2017) Clinical Pharmacokinetics and Pharmacodynamics of Lenalidomide. Clin Pharmacokinet 56:139–152.
    OpenUrl
  11. ↵
    1. Chowdhury G,
    2. Murayama N,
    3. Okada Y,
    4. Uno Y,
    5. Shimizu M,
    6. Shibata N,
    7. Guengerich FP, and
    8. Yamazaki H
    (2010) Human liver microsomal cytochrome P450 3A enzymes involved in thalidomide 5-hydroxylation and formation of a glutathione conjugate. Chem Res Toxicol 23:1018–1024.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Chowdhury G,
    2. Shibata N,
    3. Yamazaki H, and
    4. Guengerich FP
    (2014) Human cytochrome P450 oxidation of 5-hydroxythalidomide and pomalidomide, an amino analogue of thalidomide. Chem Res Toxicol 27:147–156.
    OpenUrl
  13. ↵
    1. Fujieda M,
    2. Yamazaki H,
    3. Saito T,
    4. Kiyotani K,
    5. Gyamfi MA,
    6. Sakurai M,
    7. Dosaka-Akita H,
    8. Sawamura Y,
    9. Yokota J,
    10. Kunitoh H et al.
    (2004) Evaluation of CYP2A6 genetic polymorphisms as determinants of smoking behavior and tobacco-related lung cancer risk in male Japanese smokers. Carcinogenesis 25:2451–2458.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Gordon GB,
    2. Spielberg SP,
    3. Blake DA, and
    4. Balasubramanian V
    (1981) Thalidomide teratogenesis: evidence for a toxic arene oxide metabolite. Proc Natl Acad Sci USA 78:2545–2548.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Hartiala J,
    2. Bennett BJ,
    3. Tang WH,
    4. Wang Z,
    5. Stewart AF,
    6. Roberts R,
    7. McPherson R,
    8. Lusis AJ,
    9. Hazen SL, and
    10. Allayee H
    ; CARDIoGRAM Consortium (2014) Comparative genome-wide association studies in mice and humans for trimethylamine N-oxide, a proatherogenic metabolite of choline and L-carnitine. Arterioscler Thromb Vasc Biol 34:1307–1313.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Hashizume T,
    2. Imaoka S,
    3. Mise M,
    4. Terauchi Y,
    5. Fujii T,
    6. Miyazaki H,
    7. Kamataki T, and
    8. Funae Y
    (2002) Involvement of CYP2J2 and CYP4F12 in the metabolism of ebastine in human intestinal microsomes. J Pharmacol Exp Ther 300:298–304.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Hickman D,
    2. Wang JP,
    3. Wang Y, and
    4. Unadkat JD
    (1998) Evaluation of the selectivity of In vitro probes and suitability of organic solvents for the measurement of human cytochrome P450 monooxygenase activities. Drug Metab Dispos 26:207–215.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Huang YJ,
    2. Liao JF, and
    3. Tsai TH
    (2005) Concurrent determination of thalidomide in rat blood, brain and bile using multiple microdialysis coupled to liquid chromatography. Biomed Chromatogr 19:488–493.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Hucker HB,
    2. Miller JK,
    3. Hochberg A,
    4. Brobyn RD,
    5. Riordan FH, and
    6. Calesnick B
    (1967) Studies on the absorption, excretion and metabolism of dimethylsulfoxide (DMSO) in man. J Pharmacol Exp Ther 155:309–317.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Humbert JA,
    2. Hammond KB, and
    3. Hathaway WE
    (1970) Trimethylaminuria: the fish-odour syndrome. Lancet 2:770–771.
    OpenUrlPubMed
  21. ↵
    1. Hutzler JM,
    2. Balogh LM,
    3. Zientek M,
    4. Kumar V, and
    5. Tracy TS
    (2009) Mechanism-based inactivation of cytochrome P450 2C9 by tienilic acid and (+/−)-suprofen: a comparison of kinetics and probe substrate selection. Drug Metab Dispos 37:59–65.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Ishikawa H,
    2. Mutoh M,
    3. Sato Y,
    4. Doyama H,
    5. Tajika M,
    6. Tanaka S,
    7. Horimatsu T,
    8. Takeuchi Y,
    9. Kashida H,
    10. Tashiro J et al.
    (2021) Chemoprevention with low-dose aspirin, mesalazine, or both in patients with familial adenomatous polyposis without previous colectomy (J-FAPP Study IV): a multicentre, double-blind, randomised, two-by-two factorial design trial. Lancet Gastroenterol Hepatol 6:474–481.
    OpenUrl
  23. ↵
    1. Ishikawa H,
    2. Mutoh M,
    3. Suzuki S,
    4. Tokudome S,
    5. Saida Y,
    6. Abe T,
    7. Okamura S,
    8. Tajika M,
    9. Joh T,
    10. Tanaka S et al.
    (2014) The preventive effects of low-dose enteric-coated aspirin tablets on the development of colorectal tumours in Asian patients: a randomised trial. Gut 63:1755–1759.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Ito K,
    2. Iwatsubo T,
    3. Kanamitsu S,
    4. Ueda K,
    5. Suzuki H, and
    6. Sugiyama Y
    (1998) Prediction of pharmacokinetic alterations caused by drug-drug interactions: metabolic interaction in the liver. Pharmacol Rev 50:387–412.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Ito T,
    2. Ando H,
    3. Suzuki T,
    4. Ogura T,
    5. Hotta K,
    6. Imamura Y,
    7. Yamaguchi Y, and
    8. Handa H
    (2010) Identification of a primary target of thalidomide teratogenicity. Science 327:1345–1350.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Jones KC and
    2. Ballou DP
    (1986) Reactions of the 4a-hydroperoxide of liver microsomal flavin-containing monooxygenase with nucleophilic and electrophilic substrates. J Biol Chem 261:2553–2559.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Kazuki Y,
    2. Akita M,
    3. Kobayashi K,
    4. Osaki M,
    5. Satoh D,
    6. Ohta R,
    7. Abe S,
    8. Takehara S,
    9. Kazuki K,
    10. Yamazaki H et al.
    (2016) Thalidomide-induced limb abnormalities in a humanized CYP3A mouse model. Sci Rep 6:21419.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Kim JH and
    2. Scialli AR
    (2011) Thalidomide: the tragedy of birth defects and the effective treatment of disease. Toxicol Sci 122:1–6.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Kim Y and
    2. Schmidt-Wolf IG
    (2015) Lenalidomide in multiple myeloma. Expert Rev Anticancer Ther 15:491–497.
    OpenUrl
  30. ↵
    1. Kiyotani K,
    2. Yamazaki H,
    3. Fujieda M,
    4. Iwano S,
    5. Matsumura K,
    6. Satarug S,
    7. Ujjin P,
    8. Shimada T,
    9. Guengerich FP,
    10. Parkinson A et al.
    (2003) Decreased coumarin 7-hydroxylase activities and CYP2A6 expression levels in humans caused by genetic polymorphism in CYP2A6 promoter region (CYP2A6*9). Pharmacogenetics 13:689–695.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Koyanagi T,
    2. Nakanishi Y,
    3. Murayama N,
    4. Yamaura Y,
    5. Ikeda K,
    6. Yano K,
    7. Uehara S,
    8. Utoh M,
    9. Kim S,
    10. Uno Y et al.
    (2015) Age-related changes of hepatic clearances of cytochrome P450 probes, midazolam and R-/S-warfarin in combination with caffeine, omeprazole and metoprolol in cynomolgus monkeys using in vitro-in vivo correlation. Xenobiotica 45:312–321.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Krueger SK and
    2. Williams DE
    (2005) Mammalian flavin-containing monooxygenases: structure/function, genetic polymorphisms and role in drug metabolism. Pharmacol Ther 106:357–387.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Kuwagata M,
    2. Hasegawa T,
    3. Takashima H,
    4. Shimizu M,
    5. Kitajima S, and
    6. Yamazaki H
    (2021) Pharmacokinetics of primary metabolites 5-hydroxythalidomide and 5′-hydroxythalidomide formed after oral administration of thalidomide in the rabbit, a thalidomide-sensitive species. J Toxicol Sci 46:553–560.
    OpenUrl
  34. ↵
    1. Lake BG
    (1999) Coumarin metabolism, toxicity and carcinogenicity: relevance for human risk assessment. Food Chem Toxicol 37:423–453.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Lopez-Girona A,
    2. Mendy D,
    3. Ito T,
    4. Miller K,
    5. Gandhi AK,
    6. Kang J,
    7. Karasawa S,
    8. Carmel G,
    9. Jackson P,
    10. Abbasian M et al.
    (2012) Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia 26:2326–2335.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Lu J,
    2. Helsby N,
    3. Palmer BD,
    4. Tingle M,
    5. Baguley BC,
    6. Kestell P, and
    7. Ching LM
    (2004) Metabolism of thalidomide in liver microsomes of mice, rabbits, and humans. J Pharmacol Exp Ther 310:571–577.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Masters BS and
    2. Ziegler DM
    (1971) The distinct nature and function of NADPH-cytochrome c reductase and the NADPH-dependent mixed-function amine oxidase of porcine liver microsomes. Arch Biochem Biophys 145:358–364.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Miura T,
    2. Uehara S,
    3. Shimizu M,
    4. Murayama N,
    5. Suemizu H, and
    6. Yamazaki H
    (2021a) Roles of human cytochrome P450 1A2 in coumarin 3,4-epoxidation mediated by untreated hepatocytes and by those metabolically inactivated with furafylline in previously transplanted chimeric mice. J Toxicol Sci 46:525–530.
    OpenUrl
  39. ↵
    1. Miura T,
    2. Uehara S,
    3. Shimizu M,
    4. Murayama N,
    5. Utoh M,
    6. Suemizu H, and
    7. Yamazaki H
    (2020) Different Roles of Human Cytochrome P450 2C9 and 3A Enzymes in Diclofenac 4′- and 5-Hydroxylations Mediated by Metabolically Inactivated Human Hepatocytes in Previously Transplanted Chimeric Mice. Chem Res Toxicol 33:634–639.
    OpenUrl
  40. ↵
    1. Miura T,
    2. Uehara S,
    3. Shimizu M,
    4. Suemizu H, and
    5. Yamazaki H
    (2021b) Pharmacokinetics of primary oxidative metabolites of thalidomide in rats and in chimeric mice humanized with different human hepatocytes. J Toxicol Sci 46:311–317.
    OpenUrl
  41. ↵
    1. Mori Y,
    2. Yamazaki H,
    3. Toyoshi K,
    4. Emi Y,
    5. Uchida K,
    6. Tsutsumi M, and
    7. Konishi Y
    (1985) Inhibitory effect of organic solvents on the mutagenicity of N-nitrosodialkylamines in Salmonella. Mutat Res 142:153–158.
    OpenUrlPubMed
  42. ↵
    1. Murayama N,
    2. Kazuki Y,
    3. Satoh D,
    4. Arata K,
    5. Harada T,
    6. Shibata N,
    7. Guengerich FP, and
    8. Yamazaki H
    (2017) Induction of human cytochrome P450 3A enzymes in cultured placental cells by thalidomide and relevance to bioactivation and toxicity. J Toxicol Sci 42:343–348.
    OpenUrl
  43. ↵
    1. Murayama N,
    2. Suemizu H,
    3. Uehara S,
    4. Kusama T,
    5. Mitsui M,
    6. Kamiya Y,
    7. Shimizu M,
    8. Guengerich FP, and
    9. Yamazaki H
    (2018a) Association of pharmacokinetic profiles of lenalidomide in human plasma simulated using pharmacokinetic data in humanized-liver mice with liver toxicity detected by human serum albumin RNA. J Toxicol Sci 43:369–375.
    OpenUrl
  44. ↵
    1. Murayama N,
    2. van Beuningen R,
    3. Suemizu H,
    4. Guillouzo CG,
    5. Shibata N,
    6. Yajima K,
    7. Utoh M,
    8. Shimizu M,
    9. Chesné C,
    10. Nakamura M et al.
    (2014) Thalidomide increases human hepatic cytochrome P450 3A enzymes by direct activation of the pregnane X receptor. Chem Res Toxicol 27:304–308.
    OpenUrl
  45. ↵
    1. Murayama N,
    2. Yajima K,
    3. Hikawa M,
    4. Shimura K,
    5. Ishii Y,
    6. Takada M,
    7. Uno Y,
    8. Utoh M,
    9. Iwasaki K, and
    10. Yamazaki H
    (2018b) Assessment of multiple cytochrome P450 activities in metabolically inactivated human liver microsomes and roles of P450 2C isoforms in reaction phenotyping studies. Biopharm Drug Dispos 39:116–121.
    OpenUrl
  46. ↵
    1. Murayama N and
    2. Yamazaki H
    (2021) Metabolic activation and deactivation of dietary-derived coumarin mediated by cytochrome P450 enzymes in rat and human liver preparations. J Toxicol Sci 46:371–378.
    OpenUrl
  47. ↵
    1. Muscal JA,
    2. Sun Y,
    3. Nuchtern JG,
    4. Dauser RC,
    5. McGuffey LH,
    6. Gibson BW, and
    7. Berg SL
    (2012) Plasma and cerebrospinal fluid pharmacokinetics of thalidomide and lenalidomide in nonhuman primates. Cancer Chemother Pharmacol 69:943–947.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Nagashima S,
    2. Shimizu M,
    3. Yano H,
    4. Murayama N,
    5. Kumai T,
    6. Kobayashi S,
    7. Guengerich FP, and
    8. Yamazaki H
    (2009) Inter-individual variation in flavin-containing monooxygenase 3 in livers from Japanese: correlation with hepatic transcription factors. Drug Metab Pharmacokinet 24:218–225.
    OpenUrlPubMed
  49. ↵
    1. Nakamaru Y,
    2. Hayashi Y,
    3. Ikegawa R,
    4. Kinoshita S,
    5. Perez Madera B,
    6. Gunput D,
    7. Kawaguchi A,
    8. Davies M,
    9. Mair S,
    10. Yamazaki H et al.
    (2014) Metabolism and disposition of the dipeptidyl peptidase IV inhibitor teneligliptin in humans. Xenobiotica 44:242–253.
    OpenUrl
  50. ↵
    1. Nakamura K,
    2. Matsuzawa N,
    3. Ohmori S,
    4. Ando Y,
    5. Yamazaki H, and
    6. Matsunaga T
    (2013) Clinical evidence of pharmacokinetic changes in thalidomide therapy. Drug Metab Pharmacokinet 28:38–43.
    OpenUrl
  51. ↵
    1. Nishiyama S,
    2. Suemizu H,
    3. Shibata N,
    4. Guengerich FP, and
    5. Yamazaki H
    (2015) Simulation of Human Plasma Concentrations of Thalidomide and Primary 5-Hydroxylated Metabolites Explored with Pharmacokinetic Data in Humanized TK-NOG Mice. Chem Res Toxicol 28:2088–2090.
    OpenUrl
  52. ↵
    1. Notsu Y,
    2. Shimizu M,
    3. Sasaki T,
    4. Nakano A,
    5. Ota M,
    6. Yoshida S, and
    7. Yamazaki H
    (2020) Simple pharmacokinetic models accounting for drug monitoring results of atomoxetine and its 4-hydroxylated metabolites in Japanese pediatric patients genotyped for cytochrome P450 2D6. Drug Metab Pharmacokinet 35:191–200.
    OpenUrl
  53. ↵
    1. Obach RS
    (2022) Linezolid Metabolism is Catalyzed by CYP2J2, CYP4F2 and CYP1B1. Drug Metab Dispos 50:413–421.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    1. Okada Y,
    2. Murayama N,
    3. Yanagida C,
    4. Shimizu M,
    5. Guengerich FP, and
    6. Yamazaki H
    (2009) Drug interactions of thalidomide with midazolam and cyclosporine A: heterotropic cooperativity of human cytochrome P450 3A5. Drug Metab Dispos 37:18–23.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Omura T and
    2. Sato R
    (1962) A new cytochrome in liver microsomes. J Biol Chem 237:1375–1376.
    OpenUrlPubMed
  56. ↵
    1. Ota M,
    2. Shimizu M,
    3. Kamiya Y,
    4. Emoto C,
    5. Fukuda T, and
    6. Yamazaki H
    (2019) Adult and infant pharmacokinetic profiling of dihydrocodeine using physiologically based pharmacokinetic modeling. Biopharm Drug Dispos 40:350–357.
    OpenUrl
  57. ↵
    1. Palumbo A,
    2. Facon T,
    3. Sonneveld P,
    4. Bladè J,
    5. Offidani M,
    6. Gay F,
    7. Moreau P,
    8. Waage A,
    9. Spencer A,
    10. Ludwig H et al.
    (2008) Thalidomide for treatment of multiple myeloma: 10 years later. Blood 111:3968–3977.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    1. Parikh SJ,
    2. Evans CM,
    3. Obi JO,
    4. Zhang Q,
    5. Maekawa K,
    6. Glass KC, and
    7. Shah MB
    (2020) Structure of Cytochrome P450 2C9*2 in Complex with Losartan: Insights into the Effect of Genetic Polymorphism. Mol Pharmacol 98:529–539.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    1. Parmentier Y,
    2. Pothier C,
    3. Hewitt N,
    4. Vincent L,
    5. Caradec F,
    6. Liu J,
    7. Lin F,
    8. Trancart MM,
    9. Guillet F,
    10. Bouaita B et al.
    (2019) Direct and quantitative evaluation of the major human CYP contribution (fmCYP) to drug clearance using the in vitro Silensomes™ model. Xenobiotica 49:22–35.
    OpenUrl
  60. ↵
    1. Phillips IR and
    2. Shephard EA
    (2020) Flavin-containing monooxygenase 3 (FMO3): genetic variants and their consequences for drug metabolism and disease. Xenobiotica 50:19–33.
    OpenUrlCrossRefPubMed
  61. ↵
    1. Poulsen LL and
    2. Ziegler DM
    (1979) The liver microsomal FAD-containing monooxygenase. Spectral characterization and kinetic studies. J Biol Chem 254:6449–6455.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Racha JK,
    2. Rettie AE, and
    3. Kunze KL
    (1998) Mechanism-based inactivation of human cytochrome P450 1A2 by furafylline: detection of a 1:1 adduct to protein and evidence for the formation of a novel imidazomethide intermediate. Biochemistry 37:7407–7419.
    OpenUrlCrossRefPubMed
  63. ↵
    1. Rendic S and
    2. Guengerich FP
    (2010) Update information on drug metabolism systems--2009, part II: summary of information on the effects of diseases and environmental factors on human cytochrome P450 (CYP) enzymes and transporters. Curr Drug Metab 11:4–84.
    OpenUrlCrossRefPubMed
  64. ↵
    1. Rendic S and
    2. Guengerich FP
    (2015) Survey of Human Oxidoreductases and Cytochrome P450 Enzymes Involved in the Metabolism of Xenobiotic and Natural Chemicals. Chem Res Toxicol 28:38–42.
    OpenUrlCrossRefPubMed
  65. ↵
    1. Rendic S,
    2. Crouch RD, and
    3. Guengerich FP
    (2022) Non-P450 enzymes catalyzing oxidation-reduction of endogenous compounds and xenobiotics: Flavin-containing monooxygenase (FMO), monoamine oxidase (MAO), aldehyde oxidase (AO), and NADPH-quinone oxidoreductase (NQO). Arch Toxicol (in press).
  66. ↵
    1. Rietjens IM,
    2. Punt A,
    3. Schilter B,
    4. Scholz G,
    5. Delatour T, and
    6. van Bladeren PJ
    (2010) In silico methods for physiologically based biokinetic models describing bioactivation and detoxification of coumarin and estragole: implications for risk assessment. Mol Nutr Food Res 54:195–207.
    OpenUrlCrossRefPubMed
  67. ↵
    1. Rozewski DM,
    2. Herman SE,
    3. Towns WH 2nd.,
    4. Mahoney E,
    5. Stefanovski MR,
    6. Shin JD,
    7. Yang X,
    8. Gao Y,
    9. Li X,
    10. Jarjoura D et al.
    (2012) Pharmacokinetics and tissue disposition of lenalidomide in mice. AAPS J 14:872–882.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Shephard EA,
    2. Treacy EP, and
    3. Phillips IR
    (2012) Clinical utility gene card for: trimethylaminuria. Eur J Hum Genet 20:e1–e5.
    OpenUrl
  69. ↵
    1. Shimada T,
    2. Mimura M,
    3. Inoue K,
    4. Nakamura S,
    5. Oda H,
    6. Ohmori S, and
    7. Yamazaki H
    (1997) Cytochrome P450-dependent drug oxidation activities in liver microsomes of various animal species including rats, guinea pigs, dogs, monkeys, and humans. Arch Toxicol 71:401–408.
    OpenUrlCrossRefPubMed
  70. ↵
    1. Shimada T,
    2. Takenaka S,
    3. Kakimoto K,
    4. Murayama N,
    5. Lim YR,
    6. Kim D,
    7. Foroozesh MK,
    8. Yamazaki H,
    9. Guengerich FP, and
    10. Komori M
    (2016) Structure-function studies of naphthalene, phenanthrene, biphenyl, and their derivatives in interaction with and oxidation by cytochromes P450 2A13 and 2A6. Chem Res Toxicol 29:1029–1040.
    OpenUrlCrossRefPubMed
  71. ↵
    1. Shimada T,
    2. Yamazaki H,
    3. Mimura M,
    4. Inui Y, and
    5. Guengerich FP
    (1994) Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther 270:414–423.
    OpenUrlAbstract/FREE Full Text
  72. ↵
    1. Shimizu M,
    2. Iwano S,
    3. Uno Y,
    4. Uehara S,
    5. Inoue T,
    6. Murayama N,
    7. Onodera J,
    8. Sasaki E, and
    9. Yamazaki H
    (2014) Qualitative de novo analysis of full length cDNA and quantitative analysis of gene expression for common marmoset (Callithrix jacchus) transcriptomes using parallel long-read technology and short-read sequencing. PLoS One 9:e100936.
    OpenUrlCrossRefPubMed
  73. ↵
    1. Shimizu M,
    2. Mizugaki A,
    3. Koibuchi N,
    4. Sango H,
    5. Uenuma Y, and
    6. Yamazaki H
    (2021a) A series of simple detection systems for genetic variants of flavin-containing monooxygenase 3 (FMO3) with impaired function in Japanese subjects. Drug Metab Pharmacokinet 41:100420.
    OpenUrl
  74. ↵
    1. Shimizu M,
    2. Suemizu H,
    3. Mitsui M,
    4. Shibata N,
    5. Guengerich FP, and
    6. Yamazaki H
    (2017) Metabolic profiles of pomalidomide in human plasma simulated with pharmacokinetic data in control and humanized-liver mice. Xenobiotica 47:844–848.
    OpenUrl
  75. ↵
    1. Shimizu M,
    2. Uehara S,
    3. Suemizu H, and
    4. Yamazaki H
    (2021b) In vivo drug interactions of itopride and trimethylamine mediated by flavin-containing monooxygenase 3 in humanized-liver mice. Drug Metab Pharmacokinet 37:100369.
    OpenUrl
  76. ↵
    1. Shimizu M,
    2. Uno Y,
    3. Utoh M, and
    4. Yamazaki H
    (2020) Trimethylamine N-oxygenation in cynomolgus macaques genotyped for flavin-containing monooxygenase 3 (FMO3). Drug Metab Pharmacokinet 35:571–573.
    OpenUrl
  77. ↵
    1. Shimizu M,
    2. Yoda H,
    3. Nakakuki K,
    4. Saso A,
    5. Saito I,
    6. Hishinuma E,
    7. Saito S,
    8. Hiratsuka M, and
    9. Yamazaki H
    (2019) Genetic variants of flavin-containing monooxygenase 3 (FMO3) derived from Japanese subjects with the trimethylaminuria phenotype and whole-genome sequence data from a large Japanese database. Drug Metab Pharmacokinet 34:334–339.
    OpenUrl
  78. ↵
    1. Taniguchi-Takizawa T,
    2. Kato H,
    3. Shimizu M, and
    4. Yamazaki H
    (2021) Predicted Contributions of Flavin-Containing Monooxygenases to the N-oxygenation of Drug Candidates Based on Their Estimated Base Dissociation Constants. Curr Drug Metab 22:208–214.
    OpenUrl
  79. ↵
    1. Taniguchi-Takizawa T,
    2. Shimizu M,
    3. Kume T, and
    4. Yamazaki H
    (2015) Benzydamine N-oxygenation as an index for flavin-containing monooxygenase activity and benzydamine N-demethylation by cytochrome P450 enzymes in liver microsomes from rats, dogs, monkeys, and humans. Drug Metab Pharmacokinet 30:64–69.
    OpenUrlCrossRefPubMed
  80. ↵
    1. Teo SK,
    2. Sabourin PJ,
    3. O’Brien K,
    4. Kook KA, and
    5. Thomas SD
    (2000) Metabolism of thalidomide in human microsomes, cloned human cytochrome P-450 isozymes, and Hansen’s disease patients. J Biochem Mol Toxicol 14:140–147.
    OpenUrlCrossRefPubMed
  81. ↵
    1. Toda A,
    2. Uehara S,
    3. Inoue T,
    4. Utoh M,
    5. Kusama T,
    6. Shimizu M,
    7. Uno Y,
    8. Mogi M,
    9. Sasaki E, and
    10. Yamazaki H
    (2018) Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Xenobiotica 48:720–726.
    OpenUrl
  82. ↵
    1. Trapnell CB,
    2. Donahue SR,
    3. Collins JM,
    4. Flockhart DA,
    5. Thacker D, and
    6. Abernethy DR
    (1998) Thalidomide does not alter the pharmacokinetics of ethinyl estradiol and norethindrone. Clin Pharmacol Ther 64:597–602.
    OpenUrlCrossRefPubMed
  83. ↵
    1. Turpault S,
    2. Brian W,
    3. Van Horn R,
    4. Santoni A,
    5. Poitiers F,
    6. Donazzolo Y, and
    7. Boulenc X
    (2009) Pharmacokinetic assessment of a five-probe cocktail for CYPs 1A2, 2C9, 2C19, 2D6 and 3A. Br J Clin Pharmacol 68:928–935.
    OpenUrlCrossRefPubMed
  84. ↵
    1. Uehara S,
    2. Murayama N,
    3. Nakanishi Y,
    4. Zeldin DC,
    5. Yamazaki H, and
    6. Uno Y
    (2011) Immunochemical detection of cytochrome P450 enzymes in liver microsomes of 27 cynomolgus monkeys. J Pharmacol Exp Ther 339:654–661.
    OpenUrlAbstract/FREE Full Text
  85. ↵
    1. Uehara S,
    2. Higuchi Y,
    3. Yoneda N,
    4. Kawai K,
    5. Yamamoto M,
    6. Kamimura H,
    7. Iida Y,
    8. Oshimura M,
    9. Kazuki Y,
    10. Yamazaki H et al.
    (2022) An improved TK-NOG mouse as a novel platform for humanized liver that overcomes limitations in both male and female animals. Drug Metab Pharmacokinet 42:100410.
    OpenUrl
  86. ↵
    1. Uehara S,
    2. Shimizu M,
    3. Uno Y,
    4. Inoue T,
    5. Sasaki E, and
    6. Yamazaki H
    (2017) Marmoset Flavin-Containing Monooxygenase 3 in the Liver Is a Major Benzydamine and Sulindac Sulfide Oxygenase. Drug Metab Dispos 45:497–500.
    OpenUrlAbstract/FREE Full Text
  87. ↵
    1. Uehara S,
    2. Uno Y,
    3. Inoue T,
    4. Suzuki T,
    5. Utoh M,
    6. Sasaki E, and
    7. Yamazaki H
    (2016) Caffeine 7-N-demethylation and C-8-oxidation mediated by liver microsomal cytochrome P450 enzymes in common marmosets. Xenobiotica 46:573–578.
    OpenUrl
  88. ↵
    1. Uehara S,
    2. Uno Y, and
    3. Yamazaki H
    (2020) The marmoset cytochrome P450 superfamily: Sequence/phylogenetic analyses, genomic structure, and catalytic function. Biochem Pharmacol 171:113721.
    OpenUrl
  89. ↵
    1. Uno Y,
    2. Shimizu M,
    3. Ogawa Y,
    4. Makiguchi M,
    5. Kawaguchi H,
    6. Yamato O,
    7. Ishizuka M, and
    8. Yamazaki H
    (2022d) Molecular and functional characterization of flavin-containing monooxygenases in pigs, dogs, and cats. Biochem Pharmacol 202:115125
    OpenUrl
  90. ↵
    1. Uno Y,
    2. Jikuya S,
    3. Noda Y,
    4. Murayama N, and
    5. Yamazaki H
    (2022a) A Comprehensive Investigation of Dog Cytochromes P450 3A (CYP3A) Reveals A Functional Role of Newly Identified CYP3A98 in Small Intestine. Drug Metab Dispos DOI: 10.1124/dmd.121.000749 [published ahead of print].
  91. ↵
    1. Uno Y,
    2. Murayama N,
    3. Ijiri M,
    4. Kawaguchi H,
    5. Yamato O,
    6. Shiraishi M,
    7. Asano A,
    8. Teraoka H,
    9. Mizukawa H,
    10. Nakayama SMM et al.
    (2022b) Cytochrome P450 2J (CYP2J) Genes in Dogs, Cats, and Pigs Are Expressed and Encode Functional Drug-Metabolizing Enzymes. Drug Metab Dispos DOI: 10.1124/dmd.122.000930 [published ahead of print].
  92. ↵
    1. Uno Y,
    2. Uehara S, and
    3. Yamazaki H
    (2016) Utility of non-human primates in drug development: Comparison of non-human primate and human drug-metabolizing cytochrome P450 enzymes. Biochem Pharmacol 121:1–7.
    OpenUrl
  93. ↵
    1. Uno Y,
    2. Uehara S, and
    3. Yamazaki H
    (2018) Genetic polymorphisms of drug-metabolizing cytochrome P450 enzymes in cynomolgus and rhesus monkeys and common marmosets in preclinical studies for humans. Biochem Pharmacol 153:184–195.
    OpenUrl
  94. ↵
    1. Uno Y,
    2. Uehara S, and
    3. Yamazaki H
    (2022c) Drug-oxidizing and conjugating non-cytochrome P450 (non-P450) enzymes in cynomolgus monkeys and common marmosets as preclinical models for humans. Biochem Pharmacol 197:114887.
    OpenUrl
  95. ↵
    1. Utoh M,
    2. Suemizu H,
    3. Mitsui M,
    4. Kawano M,
    5. Toda A,
    6. Uehara S,
    7. Uno Y,
    8. Shimizu M,
    9. Sasaki E, and
    10. Yamazaki H
    (2016) Human plasma concentrations of cytochrome P450 probe cocktails extrapolated from pharmacokinetics in mice transplanted with human hepatocytes and from pharmacokinetics in common marmosets using physiologically based pharmacokinetic modeling. Xenobiotica 46:1049–1055.
    OpenUrl
  96. ↵
    1. Vassallo JD,
    2. Morrall SW,
    3. Fliter KL,
    4. Curry SM,
    5. Daston GP, and
    6. Lehman-McKeeman LD
    (2003) Liquid chromatographic determination of the glutathione conjugate and ring-opened metabolites formed from coumarin epoxidation. J Chromatogr B Analyt Technol Biomed Life Sci 794:257–271.
    OpenUrlPubMed
  97. ↵
    1. Warren KE,
    2. Goldman S,
    3. Pollack IF,
    4. Fangusaro J,
    5. Schaiquevich P,
    6. Stewart CF,
    7. Wallace D,
    8. Blaney SM,
    9. Packer R,
    10. Macdonald T et al.
    (2011) Phase I trial of lenalidomide in pediatric patients with recurrent, refractory, or progressive primary CNS tumors: Pediatric Brain Tumor Consortium study PBTC-018. J Clin Oncol 29:324–329.
    OpenUrlAbstract/FREE Full Text
  98. ↵
    1. Wu K,
    2. Giovannucci E,
    3. Byrne C,
    4. Platz EA,
    5. Fuchs C,
    6. Willett WC, and
    7. Sinha R
    (2006) Meat mutagens and risk of distal colon adenoma in a cohort of U.S. men. Cancer Epidemiol Biomarkers Prev 15:1120–1125.
    OpenUrlAbstract/FREE Full Text
  99. ↵
    1. Wynalda MA,
    2. Hauer MJ, and
    3. Wienkers LC
    (2000) Oxidation of the novel oxazolidinone antibiotic linezolid in human liver microsomes. Drug Metab Dispos 28:1014–1017.
    OpenUrlAbstract/FREE Full Text
  100. ↵
    1. Yamazaki H,
    2. Inui Y,
    3. Yun CH,
    4. Guengerich FP, and
    5. Shimada T
    (1992) Cytochrome P450 2E1 and 2A6 enzymes as major catalysts for metabolic activation of N-nitrosodialkylamines and tobacco-related nitrosamines in human liver microsomes. Carcinogenesis 13:1789–1794.
    OpenUrlCrossRefPubMed
  101. ↵
    1. Yamazaki H,
    2. Inoue K,
    3. Chiba K,
    4. Ozawa N,
    5. Kawai T,
    6. Suzuki Y,
    7. Goldstein JA,
    8. Guengerich FP, and
    9. Shimada T
    (1998) Comparative studies on the catalytic roles of cytochrome P450 2C9 and its Cys- and Leu-variants in the oxidation of warfarin, flurbiprofen, and diclofenac by human liver microsomes. Biochem Pharmacol 56:243–251.
    OpenUrlCrossRefPubMed
  102. ↵
    1. Yamazaki H,
    2. Johnson WW,
    3. Ueng Y-F,
    4. Shimada T, and
    5. Guengerich FP
    (1996) Lack of electron transfer from cytochrome b5 in stimulation of catalytic activities of cytochrome P450 3A4. Characterization of a reconstituted cytochrome P450 3A4/NADPH-cytochrome P450 reductase system and studies with apo-cytochrome b5. J Biol Chem 271:27438–27444.
    OpenUrlAbstract/FREE Full Text
  103. ↵
    1. Yamazaki H,
    2. Mimura M,
    3. Sugahara C, and
    4. Shimada T
    (1994) Catalytic roles of rat and human cytochrome P450 2A enzymes in testosterone 7 alpha- and coumarin 7-hydroxylations. Biochem Pharmacol 48:1524–1527.
    OpenUrlCrossRefPubMed
  104. ↵
    1. Yamazaki H,
    2. Shimada T,
    3. Martin MV, and
    4. Guengerich FP
    (2001) Stimulation of cytochrome P450 reactions by apo-cytochrome b5: evidence against transfer of heme from cytochrome P450 3A4 to apo-cytochrome b5 or heme oxygenase. J Biol Chem 276:30885–30891.
    OpenUrlAbstract/FREE Full Text
  105. ↵
    1. Yamazaki H and
    2. Shimizu M
    (2007) Genetic polymorphism of the flavin-containing monooxygenase 3 (FMO3) associated with trimethylaminuria (fish odor syndrome): observations from Japanese patients. Curr Drug Metab 8:487–491.
    OpenUrlCrossRefPubMed
  106. ↵
    1. Yamazaki H and
    2. Shimizu M
    (2013) Survey of variants of human flavin-containing monooxygenase 3 (FMO3) and their drug oxidation activities. Biochem Pharmacol 85:1588–1593.
    OpenUrlCrossRefPubMed
  107. ↵
    1. Yamazaki H,
    2. Shimizu M,
    3. Otani T,
    4. Mizugaki A,
    5. Mure K,
    6. Suzuki S, and
    7. Ishikawa H
    (2021) Effects of polymorphic cytochrome P450 2A6 genotypes on chemoprevention against colorectal tumors in single Japanese cohort using daily low-dose aspirin: insights into future personalized treatments. J Pharm Health Care Sci 7:26.
    OpenUrl
  108. ↵
    1. Yamazaki H,
    2. Suemizu H,
    3. Igaya S,
    4. Shimizu M,
    5. Shibata N,
    6. Nakamura M,
    7. Chowdhury G, and
    8. Guengerich FP
    (2011) In vivo formation of a glutathione conjugate derived from thalidomide in humanized uPA-NOG mice. Chem Res Toxicol 24:287–289.
    OpenUrlPubMed
  109. ↵
    1. Yamazaki H,
    2. Suemizu H,
    3. Kazuki Y,
    4. Oofusa K,
    5. Kuribayashi S,
    6. Shimizu M,
    7. Ninomiya S,
    8. Horie T,
    9. Shibata N, and
    10. Guengerich FP
    (2016a) Assessment of Protein Binding of 5-Hydroxythalidomide Bioactivated in Humanized Mice with Human P450 3A-Chromosome or Hepatocytes by Two-Dimensional Electrophoresis/Accelerator Mass Spectrometry. Chem Res Toxicol 29:1279–1281.
    OpenUrl
  110. ↵
    1. Yamazaki H,
    2. Suemizu H,
    3. Mitsui M,
    4. Shimizu M, and
    5. Guengerich FP
    (2016b) Combining Chimeric Mice with Humanized Liver, Mass Spectrometry, and Physiologically-Based Pharmacokinetic Modeling in Toxicology. Chem Res Toxicol 29:1903–1911.
    OpenUrl
  111. ↵
    1. Yamazaki H,
    2. Suemizu H,
    3. Murayama N,
    4. Utoh M,
    5. Shibata N,
    6. Nakamura M, and
    7. Guengerich FP
    (2013) In vivo drug interactions of the teratogen thalidomide with midazolam: heterotropic cooperativity of human cytochrome P450 in humanized TK-NOG mice. Chem Res Toxicol 26:486–489.
    OpenUrlCrossRefPubMed
  112. ↵
    1. Yamazaki H,
    2. Suemizu H,
    3. Shimizu M,
    4. Igaya S,
    5. Shibata N,
    6. Nakamura M,
    7. Chowdhury G, and
    8. Guengerich FP
    (2012) In vivo formation of dihydroxylated and glutathione conjugate metabolites derived from thalidomide and 5-Hydroxythalidomide in humanized TK-NOG mice. Chem Res Toxicol 25:274–276.
    OpenUrlCrossRefPubMed
  113. ↵
    1. Yamazaki H,
    2. Ueng YF,
    3. Shimada T, and
    4. Guengerich FP
    (1995) Roles of divalent metal ions in oxidations catalyzed by recombinant cytochrome P450 3A4 and replacement of NADPH--cytochrome P450 reductase with other flavoproteins, ferredoxin, and oxygen surrogates. Biochemistry 34:8380–8389.
    OpenUrlCrossRefPubMed
  114. ↵
    1. Youdim KA,
    2. Zayed A,
    3. Dickins M,
    4. Phipps A,
    5. Griffiths M,
    6. Darekar A,
    7. Hyland R,
    8. Fahmi O,
    9. Hurst S,
    10. Plowchalk DR et al.
    (2008) Application of CYP3A4 in vitro data to predict clinical drug-drug interactions; predictions of compounds as objects of interaction. Br J Clin Pharmacol 65:680–692.
    OpenUrlCrossRefPubMed
  115. ↵
    1. Yamazaki M,
    2. Shimizu M,
    3. Uno Y, and
    4. Yamazaki H
    (2014) Drug oxygenation activities mediated by liver microsomal flavin-containing monooxygenases 1 and 3 in humans, monkeys, rats, and minipigs. Biochem Pharmacol 90:159–165.
    OpenUrlCrossRefPubMed
  116. ↵
    1. Zeldis JB,
    2. Carter TL,
    3. Knight RD, and
    4. Hui J
    (2013) Pomalidomide is teratogenic in rats and rabbits and can be neurotoxic in humans. Proc Natl Acad Sci USA 110:E4819.
    OpenUrlFREE Full Text
  117. ↵
    1. Ziegler DM
    (1988) Flavin-containing monooxygenases: catalytic mechanism and substrate specificities. Drug Metab Rev 19:1–32.
    OpenUrlCrossRefPubMed
  118. ↵
    1. Ziegler DM
    (2002) An overview of the mechanism, substrate specificities, and structure of FMOs. Drug Metab Rev 34:503–511.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Drug Metabolism and Disposition: 51 (1)
Drug Metabolism and Disposition
Vol. 51, Issue 1
1 Jan 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Drug Metabolism & Disposition article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Species Specificity and Selection of Models for Drug Oxidations Mediated by Polymorphic Human Enzymes
(Your Name) has forwarded a page to you from Drug Metabolism & Disposition
(Your Name) thought you would be interested in this article in Drug Metabolism & Disposition.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research Article50th Anniversary Celebration Collection—Minireview

Roles of P450 and FMO Enzymes

Hiroshi Yamazaki and Makiko Shimizu
Drug Metabolism and Disposition January 1, 2023, 51 (1) 123-129; DOI: https://doi.org/10.1124/dmd.121.000742

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research Article50th Anniversary Celebration Collection—Minireview

Roles of P450 and FMO Enzymes

Hiroshi Yamazaki and Makiko Shimizu
Drug Metabolism and Disposition January 1, 2023, 51 (1) 123-129; DOI: https://doi.org/10.1124/dmd.121.000742
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Visual Overview
    • Abstract
    • Introduction
    • Brief Historical Perspective
    • Some Key Recent Advances
    • Current Challenges and Knowledge Gaps
    • Perspective on Future Directions
    • Conclusions
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • COVID-19 Infection and Vaccine Impacts on Drug Metabolism
  • Four Decades of CYP2B Research
  • Drug Metabolism Past and Future
Show more 50th Anniversary Celebration Collection—Minireview

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About DMD
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Journal of Pharmacology and Experimental Therapeutics
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-009X (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics